Clinical importance of maternal microchimerism in children



Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

Bilateral exchange with blood cells and cell-free substances
between a mother and a fetus and visa verse routinely occurs
during normal pregnancy. A term of microchimerism (MC)
implies a few number of cells (or DNA) detected in a child and/
or a mother which primarily originated in a genetically different
individual. Whereas MC might result from iatrogenic exposure
on an organism such as transplantation or transfusion, the
main source of MC is a naturally acquired condition as a
result of transplacental interactions between a mother and
a fetus during pregnancy. The present interest to MC can
be explained by many reasons. During pregnancy fetal cells
can be found in a mothers blood in prenatal diagnostics.
Moreover, investigation of fetal MC might elucidate some
complications due to pregnancy such as preeclampsia, or
give a new insight into pathogenesis of autoimmune diseases
such as rheumatoid arthritis, whose clinical manifestation
subsides during pregnancy. Furthermore, it is known at
present that MC retains for a long time after a child delivery
where it occurs both with fetal MC acquired by the woman
during pregnancy and with maternal MC in her descendants.
Investigations of long-term effects of fetal and maternal MC
is a novel, active research, its results indicate both favourable
and unfavourable long-term effects of MC.
The present article is a review of available data on maternal
or, to be more precise, maternal-fetal microchimerism and
described or experimentally proposed its consequences.

About the authors

A G Rumyantsev

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

M A Kurtser

Russian State N.I.Pirogov Medical University

Russian State N.I.Pirogov Medical University

Yu B Mareeva

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

A V Misyurin

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

S A Rumyantsev

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

A Yu Ustyugov

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

Federal State Research Center of Children Hematology, Oncology and Immunologyof the RF Ministry of Health and Social Development

References

  1. Reynolds A.G. Placental metastasis from malignant melanoma; report of a case. Obstet. Gynecol. 1955; 6: 205-9.
  2. Freedman W.L., McMahon F.J. Placental metastasis. Review of the literature and report of a case of metastatic melanoma. Obstet. Gynecol. 1960; 6: 550-60.
  3. Burlingham W.J., Grailer A.P., Heisey D.M. et al. The Effect of Tolerance to Noninherited Maternal HLA Antigens on the Survival of Renal Transplants from Sibling Donors. N. Engl. J. Med. 1998; 339: 1657-64.
  4. Claas F.H.J., Gijbels Y., Vandervelden-Demunck J., Rood J.J.V. Induction of В Cell Unresponsiveness to Noninherited Maternal HLA Antigens during Fetal Life. Science 1988; 241: 1815-7.
  5. Pollack M.S., Kirpatrick D., Kapoor D. et al. Identification by HLA typing of intrauterine derived maternal T cells in four patients with severe combined immunodeficiency. N. Engl. J. Meo. 1982; 307: 662-6.
  6. Maloney S., Smith A., Furst D.E. et al. Microchimerism of maternal origin persists into adult life. 1999; J. Clin. Invest. 104: 41-7.
  7. Geha R.S., Reinherz E. Identification of circulating maternal T and В lymphocytes in uncomplicated severe combined immunodeficiency by HLA typing of subpopulations of T cells separated by the fluorescenceactivated cell sorter and of Epstein Barr virus-derived В cell lines. J. Immunol. 1983; 130: 2493-5.
  8. Burlingham W.J., Grailer A.P., Heisey D.M. et al. The Effect of Tolerance to Noninherited Maternal HLA Antigens on the Survival of Renal Transplants from Sibling Donors. N. Engl. J. Med. 1998; 339: 1657-64.
  9. Stern M., Ruggeri L., Mancusi A. et al. Survival after T-cell depleted haploidentical stem cell transplantation is improved using the mother as donor. Blood 2008; 1: 135-45.
  10. Tenwolde S., Breedveld F.C., Devries R.R. et al. Influence of non-inherited maternal HLA antigens on occurrence of rheumatoid arthritis. Lancet 1993; 3: 200-2.
  11. Pani M.A., Vanautreve J., Vanderauwera B.J. et al. Nontransmitted maternal HLA DQ2 or DQ8 alleles and risk of Type I diabetes in offspring: the importance of foetal or post partum exposure to diabetogenic molecules. Diabetologia 2002; 45: 1340-3.
  12. Fernandes R.J., Deheer E., Bruijn J.A., Bajema I.M. Chimerism in Kidneys, Livers and Hearts of Normal Women: Implications for Transplantation Studies. Am. J. Transpl. 2005: 1495-502.
  13. Maurel M.C., Kanellopoulos-Langevin С. Heredity-Venturing beyond genetics. Biol. Reprod. 2008, 79: 2-8.
  14. Lo Y.M., Lo E.S., Watson N. et al. Two-way cell traffic between mother and fetus: biologic and clinical implications. Blood 1996; 8: 4390-5.
  15. Lo Y., Lau Т., Chan L. et al. Quantitative analysis of the bidirectional fetomaternal transfer of nucleated cells and plasma DNA. Clin. Chem. 2000; 46(9): 1301-9.
  16. Berry S.M., Hassan S.S., Russell et al. Association of maternal histocompatibility at Class II loci with maternal microchimerism in the fetus. Pediatr. Res. 2004; 56: 73-8.
  17. Portanova J.P., Kotzin B.L. Lupus-like autoimmunity in murine graft-versus-host disease. Concepts Immunopathol. 1988; 6: 119-40.
  18. Stevens A.M., Tsao B.P., Hahn B.H. et al. Maternal HLA class II compatibility in men with systemic lupus erythematosus. Arthritis Rheum. 2005; 52: 2768-73.
  19. Kaplan J., Land S. Influence of Maternal-Fetal Histocompatibility and MHC Zygosity on Maternal Microchimerism. J. Immunol. 2005; 174: 7123-8.
  20. Gotherstrom C., Johnsson A.M., Mattsson J. et al. Identificantion of maternal hematopoietic cells in a 2nd-trimester fetus. Fetal Diagn. Ther. 2005; 20: 355-8.
  21. Johnson K.L., McAundon Т.Е., Mulcahy E., Bianchi D.W. Microchimerism in a female patient with systemic lupus erythematosus. Arthritis Rheum. 2001; 44: 210-11.
  22. Srivatsa В., Srivatsa S., Johnson K.L. et al. Microchimerism of presumed fetal origin in thyroid specimens from women: a casecontrol study. Lancet 2001; 358: 2034-8.
  23. Nelson J.L., Gillespie K.M., Lambert N.C. et al. Maternal microchimerism in peripheral blood in type 1 diabetes and pancreatic islet beta cell microchimerism. PNAS USA 2007; 104: 1637-42.
  24. Khosrotehrani K., Johnson K.L., Guegan S. et al. Natural history of fetal cell microchimerism during and following murine pregnancy. J. Reprod. Immunol. 2005; 6: 1-12.
  25. Loubiere L.S., Lambert N.C., Flinn L.J. et al. Maternal microchimerism in healthy adults in lymphocytes, monocyte/ macrophages and NK cells. Lablnvestm/2006, 86: 1185-1192.
  26. Chen С.Р., Lee M.Y., Huang J.P. et al. Trafficking of Multtpotent Mesenchymal Stromal Cells from Maternal Circulation Through the Placenta Involves Vascular Endothelial Growth Factor Receptor-1 and Integrins. Stem Cells 2008; 26: 550-61.
  27. Piotrowski P., Croy B.A. Maternal cells are widely distributed in murine fetuses in utero. Biol. Reprod. 1996; 54: 1103-10.
  28. Su E.C., Johnson K.L., Tighiouart H., Bianchi D.W. Murine Maternal Cell Microchimerism: Analysis Using Real-Time PCR and in vivo Imaging. Biol. Reprod. 2008; 78: 883-7.
  29. Wrenshall L.E., Stevens E.T., Smith D.R., Miller J.D. Maternal microchimerism leads to the presence of interleukin-2 in interleukin-2 knock out mice: Implications for the role of interleukin-2 in thymic function. Cell Immuno. 2007; 245: 80-90.
  30. Surbek D.V., Gratwohl A., Holzgreve W. In utero hematopoietic stem cell transfer: current status and future strategies. Eur. J. Obstet. Gynecol. Reprod. Biol. 1999; 85: 109-15.
  31. Garbuzva-Davis S., Gografe S.J., Sanberg C.D. et al. Maternal transplantation of human umbilical cord blood cells provides prenatal therapy in Sanfilippo type В mouse model. FASEB J. 2006; 20(3): 485-7.
  32. Carrier E., Gilpin E., Lee Т.Н. et al. Microchimerism does not induce tolerance after in utero transplantation and may lead to the development of alloreactivity. J. Lab. Clin. Med. 2000; 136: 224-35.
  33. Kowalzick L., Artlett C., Thiss K. et al. Chronic graft-versushost- disease-likedermopathy in a child with CD4+cell microchimerism. Dermatology 2005; 210(1): 68-71.
  34. Lambert N.C., Erickson T.D., Yan Z. et al. Quantification of maternal microchimerism by HLA-specific real-time polymerase chain reaction: Studies of healthy women and women with scleroderma. Arthritis Rheum. 2004; 50: 906-14.
  35. Stevens A.M., Hermes H.M., Rutledge J.C. et al. Myocardialtissue- specific phenotype of maternal microchimerism in neonatal lupus congenital heart block. Lancet 2003; 362: 1617-23.
  36. Stevens A.M., Hermes H.M., Lambert N.C. et al. Maternal and sibling microchimerism in twins and triplets discordant for neonatal lupus syndrome-congenital heart block. Rheumatology 2005; 44: 187-91.
  37. Artlett С.М., Ramos R., Jiminez S.A. et al. Chimeric cells of maternal origin in juvenile idiopathic inflammatory myopathies. Lancet 2000; 356: 2155-6.
  38. Reed A.M., Picornell Y.J., Harwood A., Kredish D.W. Chimerism in children with juvenile dermatomyositis. Lancet 2000; 356: 2156-7.
  39. Artlett С.М., Miller F.W., Rider L.G. et al. Persistent maternally derived peripheral microchimerism is associated with the juvenile idiopathic inflammatory myopathies. Rheumatology 2001; 40: 1279-84.
  40. Khosrotehrani K. Mery L., Aractingi S. et al. Absence of fetal cell microchimerism in cutaneous lesions of lupus erythematosus. Ann. Rheum. Dis. 2005, 64: 159-60.
  41. Kobayashi H., Tamatani Т., Tamura Т. et al. Maternal microchimerism in biliary atresia. J. Pediatr. Surg. 2007; 42: 987-91.
  42. Regnier S., Dupin N., Danff C.L. et al. Endothelial cells in infantile haemangiomas originate from the child and not from the mother (a fluorescence in situ hybridization-based study). Br. J. Dermatol. 2007; 157: 158-60.
  43. Willer C.J., Herrera B.M., Morrison K.M.E. et al. Association between microchimerism and multiple sclerosis in Canadian twins. J. Neuroimmunol. 2006; 179: 145-51.

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2011 Eco-Vector



СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: 

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies